Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Inorg Chem ; 63(9): 4419-4428, 2024 Mar 04.
Article in English | MEDLINE | ID: mdl-38364266

ABSTRACT

The combination of photodynamic therapy and radiotherapy has given rise to a modality called radiodynamic therapy (RDT), based on reactive oxygen species-producing radiosensitizers. The production of singlet oxygen, O2(1Δg), by octahedral molybdenum (Mo6) clusters upon X-ray irradiation allows for simplification of the architecture of radiosensitizing systems. In this context, we prepared a radiosensitizing system using copper-free click chemistry between a Mo6 cluster bearing azido ligands and the homo-bifunctional linker bis-dPEG11-DBCO. The resulting compound formed nanoparticles, which featured production of O2(1Δg) and efficient cellular uptake, leading to remarkable photo- and radiotoxic effects against the prostatic adenocarcinoma TRAMP-C2 cell line. Spheroids of TRAMP-C2 cells were also used for evaluation of toxicity and phototoxicity. In vivo experiments on a mouse model demonstrated that subcutaneous injection of the nanoparticles is a safe administration mode at a dose of up to 0.08 g kg-1. The reported results confirm the relevancy of Mo6-based radiosensitizing nanosystems for RDT.


Subject(s)
Adenocarcinoma , Iodine , Photochemotherapy , Animals , Mice , Molybdenum/chemistry , Photochemotherapy/methods , Polyethylene Glycols
2.
Cell Death Dis ; 14(3): 209, 2023 03 24.
Article in English | MEDLINE | ID: mdl-36964168

ABSTRACT

While type I interferon (IFN) is best known for its key role against viral infection, accumulating preclinical and clinical data indicate that robust type I IFN production in the tumor microenvironment promotes cancer immunosurveillance and contributes to the efficacy of various antineoplastic agents, notably immunogenic cell death inducers. Here, we report that malignant blasts from patients with acute myeloid leukemia (AML) release type I IFN via a Toll-like receptor 3 (TLR3)-dependent mechanism that is not driven by treatment. While in these patients the ability of type I IFN to stimulate anticancer immune responses was abolished by immunosuppressive mechanisms elicited by malignant blasts, type I IFN turned out to exert direct cytostatic, cytotoxic and chemosensitizing activity in primary AML blasts, leukemic stem cells from AML patients and AML xenograft models. Finally, a genetic signature of type I IFN signaling was found to have independent prognostic value on relapse-free survival and overall survival in a cohort of 132 AML patients. These findings delineate a clinically relevant, therapeutically actionable and prognostically informative mechanism through which type I IFN mediates beneficial effects in patients with AML.


Subject(s)
Antineoplastic Agents , Interferon Type I , Leukemia, Myeloid, Acute , Humans , Leukemia, Myeloid, Acute/pathology , Antineoplastic Agents/therapeutic use , Treatment Outcome , Signal Transduction , Tumor Microenvironment
3.
Mol Med Rep ; 27(4)2023 04.
Article in English | MEDLINE | ID: mdl-36825563

ABSTRACT

Signal transducer and activator of transcription 3 (STAT3) signalling serves an important role in carcinogenesis and cellular senescence, and its inhibition in tumour cells represents an attractive therapeutic target. Premature cellular senescence, a process of permanent proliferative arrest of cells in response to various inducers, such as cytostatic drugs or ionizing radiation, is accompanied by morphological and secretory changes, and by altered susceptibility to chemotherapeutic agents, which can thereby complicate their eradication by cancer therapies. In the present study, the responsiveness of proliferating and docetaxel (DTX)­induced senescent cancer cells to small molecule STAT3 inhibitor Stattic and its analogues was evaluated using tumour cell lines. These agents displayed cytotoxic effects in cell viability assays on both proliferating and senescent murine TRAMP­C2 and TC­1 cells; however, senescent cells were markedly more resistant. Western blot analysis revealed that Stattic and its analogues effectively inhibited constitutive STAT3 phosphorylation in both proliferating and senescent cells. Furthermore, whether the Stattic­derived inhibitor K1836 could affect senescence induction or modulate the phenotype of senescent cells was evaluated. K1836 treatment demonstrated no effect on senescence induction by DTX. However, the K1836 compound significantly modulated secretion of certain cytokines (interleukin­6, growth­regulated oncogene α and monocyte chemoattractant protein­1). In summary, the present study demonstrated differences between proliferating and senescent tumour cells in terms of their susceptibility to STAT3 inhibitors and demonstrated the ability of the new STAT3 inhibitor K1836 to affect the secretion of essential components of the senescence­associated secretory phenotype. The present study may be useful for further development of STAT3 inhibitor­based therapy of cancer or age­related diseases.


Subject(s)
Cytokines , STAT3 Transcription Factor , Animals , Mice , Phosphorylation , STAT3 Transcription Factor/metabolism , Gene Expression , Docetaxel/pharmacology , Cytokines/metabolism , Cellular Senescence
4.
EMBO Rep ; 24(1): e54729, 2023 01 09.
Article in English | MEDLINE | ID: mdl-36341527

ABSTRACT

Chronic inflammation represents a major threat to human health since long-term systemic inflammation is known to affect distinct tissues and organs. Recently, solid evidence demonstrated that chronic inflammation affects hematopoiesis; however, how chronic inflammation affects hematopoietic stem cells (HSCs) on the mechanistic level is poorly understood. Here, we employ a mouse model of chronic multifocal osteomyelitis (CMO) to assess the effects of a spontaneously developed inflammatory condition on HSCs. We demonstrate that hematopoietic and nonhematopoietic compartments in CMO BM contribute to HSC expansion and impair their function. Remarkably, our results suggest that the typical features of murine multifocal osteomyelitis and the HSC phenotype are mechanistically decoupled. We show that the CMO environment imprints a myeloid gene signature and imposes a pro-inflammatory profile on HSCs. We identify IL-6 and the Jak/Stat3 signaling pathway as critical mediators. However, while IL-6 and Stat3 blockage reduce HSC numbers in CMO mice, only inhibition of Stat3 activity significantly rescues their fitness. Our data emphasize the detrimental effects of chronic inflammation on stem cell function, opening new venues for treatment.


Subject(s)
Inflammation , Interleukin-6 , Humans , Animals , Mice , Interleukin-6/genetics , Interleukin-6/metabolism , Inflammation/metabolism , Signal Transduction , Hematopoiesis , Hematopoietic Stem Cells/metabolism , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism
5.
Front Aging Neurosci ; 14: 1048260, 2022.
Article in English | MEDLINE | ID: mdl-36561137

ABSTRACT

To date, the most studied drug in anti-aging research is the mTOR inhibitor - rapamycin. Despite its almost perfect anti-aging profile, rapamycin exerts one significant limitation - inappropriate physicochemical properties. Therefore, we have decided to utilize virtual high-throughput screening and fragment-based design in search of novel mTOR inhibiting scaffolds with suitable physicochemical parameters. Seven lead compounds were selected from the list of obtained hits that were commercially available (4, 5, and 7) or their synthesis was feasible (1, 2, 3, and 6) and evaluated in vitro and subsequently in vivo. Of all these substances, only compound 3 demonstrated a significant cytotoxic, senolytic, and senomorphic effect on normal and cancerous cells. Further, it has been confirmed that compound 3 is a direct mTORC1 inhibitor. Last but not least, compound 3 was found to exhibit anti-SASP activity concurrently being relatively safe within the test of in vivo tolerability. All these outstanding results highlight compound 3 as a scaffold worthy of further investigation.

6.
J Mater Chem B ; 10(17): 3303-3310, 2022 05 04.
Article in English | MEDLINE | ID: mdl-35380154

ABSTRACT

X-Ray-induced photodynamic therapy represents a suitable modality for the treatment of various malignancies. It is based on the production of reactive oxygen species by radiosensitizing nanoparticles activated by X-rays. Hence, it allows overcoming the depth-penetration limitations of conventional photodynamic therapy and, at the same time, reducing the dose needed to eradicate cancer in the frame of radiotherapy treatment. The direct production of singlet oxygen by octahedral molybdenum cluster complexes upon X-ray irradiation is a promising avenue in order to simplify the architecture of radiosensitizing systems. One such complex was utilized to prepare water-stable nanoparticles using the solvent displacement method. The nanoparticles displayed intense red luminescence in aqueous media, efficiently quenched by oxygen to produce singlet oxygen, resulting in a substantial photodynamic effect under blue light irradiation. A robust radiosensitizing effect of the nanoparticles was demonstrated in vitro against TRAMP-C2 murine prostatic carcinoma cells at typical therapeutic X-ray doses. Injection of a suspension of the nanoparticles to a mouse model revealed the absence of acute toxicity as evidenced by the invariance of key physiological parameters. This study paves the way for the application of octahedral molybdenum cluster-based radiosensitizers in X-ray-induced photodynamic therapy and its translation to in vivo experiments.


Subject(s)
Carcinoma , Nanoparticles , Photochemotherapy , Prostatic Neoplasms , Radiation-Sensitizing Agents , Animals , Humans , Male , Mice , Molybdenum/pharmacology , Photochemotherapy/methods , Prostatic Neoplasms/drug therapy , Singlet Oxygen , X-Rays
7.
J Enzyme Inhib Med Chem ; 36(1): 410-424, 2021 Dec.
Article in English | MEDLINE | ID: mdl-33440995

ABSTRACT

Twelve novel analogs of STAT3 inhibitor BP-1-102 were designed and synthesised with the aim to modify hydrophobic fragments of the molecules that are important for interaction with the STAT3 SH2 domain. The cytotoxic activity of the reference and novel compounds was evaluated using several human and two mouse cancer cell lines. BP-1-102 and its two analogs emerged as effective cytotoxic agents and were further tested in additional six human and two murine cancer cell lines, in all of which they manifested the cytotoxic effect in a micromolar range. Reference compound S3I-201.1066 was found ineffective in all tested cell lines, in contrast to formerly published data. The ability of selected BP-1-102 analogs to induce apoptosis and inhibition of STAT3 receptor-mediated phosphorylation was confirmed. The structure-activity relationship confirmed a demand for two hydrophobic substituents, i.e. the pentafluorophenyl moiety and another spatially bulky moiety, for effective cytotoxic activity and STAT3 inhibition.


Subject(s)
Aminosalicylic Acids/pharmacology , Antineoplastic Agents/pharmacology , Drug Design , STAT3 Transcription Factor/antagonists & inhibitors , Sulfonamides/pharmacology , Aminosalicylic Acids/chemical synthesis , Aminosalicylic Acids/chemistry , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Humans , Hydrophobic and Hydrophilic Interactions , Mice , Mice, Inbred C57BL , Molecular Structure , Phosphorylation/drug effects , STAT3 Transcription Factor/metabolism , Structure-Activity Relationship , Sulfonamides/chemical synthesis , Sulfonamides/chemistry
8.
Haematologica ; 105(7): 1868-1878, 2020 07.
Article in English | MEDLINE | ID: mdl-31582537

ABSTRACT

In some settings, cancer cells responding to treatment undergo an immunogenic form of cell death that is associated with the abundant emission of danger signals in the form of damage-associated molecular patterns. Accumulating preclinical and clinical evidence indicates that danger signals play a crucial role in the (re-)activation of antitumor immune responses in vivo, thus having a major impact on patient prognosis. We have previously demonstrated that the presence of calreticulin on the surface of malignant blasts is a positive prognostic biomarker for patients with acute myeloid leukemia (AML). Calreticulin exposure not only correlated with enhanced T-cell-dependent antitumor immunity in this setting but also affected the number of circulating natural killer (NK) cells upon restoration of normal hematopoiesis. Here, we report that calreticulin exposure on malignant blasts is associated with enhanced NK cell cytotoxic and secretory functions, both in AML patients and in vivo in mice. The ability of calreticulin to stimulate NK-cells relies on CD11c+CD14high cells that, upon exposure to CRT, express higher levels of IL-15Rα, maturation markers (CD86 and HLA-DR) and CCR7. CRT exposure on malignant blasts also correlates with the upregulation of genes coding for type I interferon. This suggests that CD11c+CD14high cells have increased capacity to migrate to secondary lymphoid organs, where can efficiently deliver stimulatory signals (IL-15Rα/IL-15) to NK cells. These findings delineate a multipronged, clinically relevant mechanism whereby surface-exposed calreticulin favors NK-cell activation in AML patients.


Subject(s)
Calreticulin , Leukemia, Myeloid, Acute , Animals , Calreticulin/genetics , Calreticulin/metabolism , Cytotoxicity, Immunologic , Humans , Interleukin-15 , Killer Cells, Natural , Leukemia, Myeloid, Acute/therapy , Lymphocyte Activation , Mice
9.
Int J Oncol ; 53(5): 1997-2009, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30226595

ABSTRACT

Cellular senescence is the process of the permanent proliferative arrest of cells in response to various inducers. It is accompanied by typical morphological changes, in addition to the secretion of bioactive molecules, including proinflammatory cytokines and chemokines [known as the senescence-associated secretory phenotype (SASP)]. Thus, senescent cells may affect their local environment and induce a so-called 'bystander' senescence through the state of SASP. The phenotypes of senescent cells are determined by the type of agent inducing cellular stress and the cell lineages. To characterise the phenotypes of senescent cancer cells, two murine cell lines were employed in the present study: TC-1 and B16F10 (B16) cells. Two distinct senescence inductors were used: Chemotherapeutic agent docetaxel (DTX) and a combination of immunomodulatory cytokines, including interferon Î³ (IFNγ) and tumour necrosis factor α (TNFα). It was demonstrated that DTX induced senescence in TC-1 and B16 tumour cell lines, which was demonstrated by growth arrest, positive ß-galactosidase staining, increased p21Waf1 (p21) expression and the typical SASP capable of inducing a 'bystander' senescence. By contrast, treatment with a combination of T helper cell 1 cytokines, IFNγ and TNFα, induced proliferation arrest only in B16 cells. Despite the presence of certain characteristic features resembling senescent cells (proliferation arrest, morphological changes and increased p21 expression), these cells were able to form tumours in vivo and started to proliferate upon cytokine withdrawal. In addition, B16 cells were not able to induce a 'bystander' senescence. In summary, the present study described cell line- and treatment-associated differences in the phenotypes of senescent cells that may be relevant in optimization of cancer chemo- and immunotherapy.


Subject(s)
Antineoplastic Agents/pharmacology , Bystander Effect/immunology , Cellular Senescence/immunology , Docetaxel/pharmacology , Interferon-gamma/metabolism , Neoplasms/immunology , Tumor Necrosis Factor-alpha/metabolism , Animals , Antineoplastic Agents/therapeutic use , Bystander Effect/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cellular Senescence/drug effects , Disease Models, Animal , Docetaxel/therapeutic use , Humans , Interferon-gamma/immunology , Male , Mice , Mice, Inbred C57BL , Neoplasms/drug therapy , Neoplasms/pathology , Phenotype , Tumor Necrosis Factor-alpha/immunology
10.
Oncol Lett ; 15(3): 3594-3601, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29467880

ABSTRACT

The present study aimed to elucidate the role of cluster of differentiation (CD)8+, CD4+, natural killer (NK), and myeloid (CD11b+) cells in the course of the growth and rejection of experimental major histocompatibility complex (MHC) class I-deficient, HPV16 E6/E7-associated TC-1/A9 tumors in mice. Stable mouse lines (F30) generated by inbreeding of Balb/c and C57BL/6 strains, which were characterized by H-2Db+d-NK1.1neg (B6-neg) and H-2Db-d+NK1.1high (Balb-high) phenotypes, were used for the present study. The novel strains spontaneously regressed tumors in 70-90% of cases. Ex vivo histological analysis of the tumor microenvironment in cryosections showed an indirect correlation between the growth of the transplanted tumor (progressor vs. regressor mice) and the proportion of immunocompetent cell infiltration in the tumors. The regressor mice exhibited a higher infiltration of tumors with CD4+ and CD8+ cells, and in Balb-high with NK cells as well, compared with the progressors. All tumor transplants also indicated a huge infiltration of CD11b+ cells, but this infiltration was not dependent on the stage of the TC-1/A9 tumor development. Depletion of individual cell subpopulations in vivo exhibited different effects on the tumor development in the two strains. Elimination of CD8-positive cells enhanced growth of TC-1/A9 tumor transplants in both hybrid stains, whereas CD4+ cell depletion affected rejection of TC-1/A9 tumors in the B6-neg mice only. Depletion of NK cells with anti-asialo GM1 antibody in the Balb-high strain led to enhancement of tumor growth, which was more pronounced after depletion of the NK1.1+ subpopulation. On the other hand, depletion of NK cells with anti-asialo GM1 in B6-neg mice did not affect the regression of TC-1/A9 tumor transplants, but increased the CD11b+ cell infiltration. In summary, these results indicate that co-operation of particular subsets of immunocompetent cells is essential for the rejection of TC-1/A9 tumor transplants. In B6-neg mice, the co-operative action of CD8+ and CD4+ cells is required, whereas in Balb-high mice, the synergy of CD8+ and NK1.1+ cells is of major importance.

11.
Oncoimmunology ; 6(12): e1362528, 2017.
Article in English | MEDLINE | ID: mdl-29209567

ABSTRACT

Dendritic cell (DC)-based vaccines pulsed with high hydrostatic pressure (HHP)-inactivated tumor cells have recently been shown to be a promising tool for prostate cancer chemoimmunotherapy. In this study, DC-based vaccines, both pulsed and unpulsed, were as effective as docetaxel (DTX) in reducing prostate tumors in the orthotopic transgenic adenocarcinoma of the mouse prostate (TRAMP) model. However, we did not observe any additive or synergic effects of chemoimmunotherapy on the tumor growth, while only the combination of DTX and pulsed dendritic cells resulted in significantly lower proliferation detected by Ki67 staining in histological samples. The DC-based vaccine pulsed with HHP-treated tumor cells was also combined with another type of cytostatic, cyclophosphamide, with similar results. In another clinically relevant setting, minimal residual tumor disease after surgery, administration of DC-based vaccines after the surgery of poorly immunogenic transplanted TRAMP-C2, as well as in immunogenic TC-1 tumors, reduced the growth of tumor recurrences. To identify the effector cell populations after DC vaccine application, mice were twice immunized with both pulsed and unpulsed DC vaccine, and the cytotoxicity of the spleen cells populations was tested. The effector cell subpopulations were defined as CD4+ and NK1.1+, which suggests rather unspecific therapeutic effects of the DC-based vaccines in our settings. Taken together, our data demonstrate that DC-based vaccines represent a rational tool for the treatment of human prostate cancer.

12.
Oncotarget ; 7(34): 54952-54964, 2016 Aug 23.
Article in English | MEDLINE | ID: mdl-27448982

ABSTRACT

Standard-of-care chemo- or radio-therapy can induce, besides tumor cell death, also tumor cell senescence. While senescence is considered to be a principal barrier against tumorigenesis, senescent cells can survive in the organism for protracted periods of time and they can promote tumor development. Based on this emerging concept, we hypothesized that elimination of such potentially cancer-promoting senescent cells could offer a therapeutic benefit. To assess this possibility, here we first show that tumor growth of proliferating mouse TC-1 HPV-16-associated cancer cells in syngeneic mice becomes accelerated by co-administration of TC-1 or TRAMP-C2 prostate cancer cells made senescent by pre-treatment with the anti-cancer drug docetaxel, or lethally irradiated. Phenotypic analyses of tumor-explanted cells indicated that the observed acceleration of tumor growth was attributable to a protumorigenic environment created by the co-injected senescent and proliferating cancer cells rather than to escape of the docetaxel-treated cells from senescence. Notably, accelerated tumor growth was effectively inhibited by cell immunotherapy using irradiated TC-1 cells engineered to produce interleukin IL-12. Collectively, our data document that immunotherapy, such as the IL-12 treatment, can provide an effective strategy for elimination of the detrimental effects caused by bystander senescent tumor cells in vivo.


Subject(s)
Cellular Senescence/drug effects , Immunotherapy, Adoptive/methods , Interleukin-12/pharmacology , Neoplasms, Experimental/therapy , Taxoids/pharmacology , Tumor Burden/drug effects , Animals , Antineoplastic Agents/pharmacology , Bystander Effect/drug effects , Cell Line, Tumor , Combined Modality Therapy , Cytokines/genetics , Cytokines/metabolism , Docetaxel , Interleukin-12/biosynthesis , Male , Mice, Inbred C57BL , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Time Factors
13.
Int J Oncol ; 49(2): 763-72, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27279019

ABSTRACT

To elucidate the immunological mechanisms critical for tumor progression, we bred novel mouse strains, different in the NKC and H-2D domains. We used inbreeding to generate hybrids of Balb/c and C57BL/6 of stable H-2Db+d-NK1.1neg and H-2Db-d+NK1.1high phenotypes. We analyzed the growth of three established MHC class I-deficient tumor cell lines: TC-1/A9 tumor (HPV-associated) and B16F10 melanoma, both syngeneic to C57BL/6, and the MCB8 (3-methycholanthrene-induced tumor) syngeneic to Balb/c. Furthermore, we induced colorectal carcinoma by azoxymethane-DSS treatment to test the susceptibility to chemically-induced primary cancer. We found that the novel strains spontaneously regressed the tumor transplants syngeneic to both Balb/c (MCB8) and C57BL/6 (B16F10 and TC-1/A9) mice. The H2-Db+d-NK1.1neg, but not the H2-Db-d+NK1.1high strain was also highly resistant to chemically-induced colorectal cancer in comparison to the parental mice. The immune changes during TC-1/A9 cancer development involved an increase of the NK cell distribution in the peripheral blood and spleen along with higher expression of NKG2D activation antigen; this was in correlation with the time-dependent rise of cytotoxic activity in comparison to C57BL/6 mice. The TC-1/A9 cancer regression was accompanied by higher proportion of B cells in the spleen and B220+/CD86+ activated antigen-presenting B cells distributed in the lymphoid organs, as well as in the periphery. The changes in the T-cell population were represented mainly by the prevalence of T helper cells reflected by grown CD4/CD8 ratio, most prominent in the b+d-NK1.1neg strain. The results of the present study imply usefulness of the two novel mouse strains as an experimental model for further studies of tumor resistance mechanisms.


Subject(s)
Histocompatibility Antigens Class I/immunology , Killer Cells, Natural/immunology , Neoplasms, Experimental/genetics , Animals , Female , Gene Expression Regulation, Neoplastic/genetics , Histocompatibility Antigens Class I/genetics , Humans , Killer Cells, Natural/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , NK Cell Lectin-Like Receptor Subfamily K/biosynthesis , NK Cell Lectin-Like Receptor Subfamily K/immunology , Neoplasms, Experimental/immunology , Neoplasms, Experimental/pathology
14.
Int J Oncol ; 48(3): 953-64, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26718011

ABSTRACT

High hydrostatic pressure (HHP) has been shown to induce immunogenic cell death of cancer cells, facilitating their uptake by dendritic cells (DC) and subsequent presentation of tumor antigens. In the present study, we demonstrated immunogenicity of the HHP-treated tumor cells in mice. HHP was able to induce immunogenic cell death of both TC-1 and TRAMP-C2 tumor cells, representing murine models for human papilloma virus-associated tumors and prostate cancer, respectively. HHP-treated cells induced stronger immune responses in mice immunized with these tumor cells, documented by higher spleen cell cytotoxicity and increased IFNγ production as compared to irradiated tumor cells, accompanied by suppression of tumor growth in vivo in the case of TC-1 tumors, but not TRAMP-C2 tumors. Furthermore, HHP-treated cells were used for DC-based vaccine antigen pulsing. DC co-cultured with HHP-treated tumor cells and matured by a TLR 9 agonist exhibited higher cell surface expression of maturation markers and production of IL-12 and other cytokines, as compared to the DC pulsed with irradiated tumor cells. Immunization with DC cell-based vaccines pulsed with HHP-treated tumor cells induced high immune responses, detected by increased spleen cell cytotoxicity and elevated IFNγ production. The DC-based vaccine pulsed with HHP-treated tumor cells combined with docetaxel chemotherapy significantly inhibited growth of both TC-1 and TRAMP-C2 tumors. Our results indicate that DC-based vaccines pulsed with HHP-inactivated tumor cells can be a suitable tool for chemoimmunotherapy, particularly with regard to the findings that poorly immunogenic TRAMP-C2 tumors were susceptible to this treatment modality.


Subject(s)
Antineoplastic Agents/administration & dosage , Dendritic Cells/cytology , Neoplasms, Experimental/therapy , Papillomavirus Infections/therapy , Prostatic Neoplasms/therapy , Taxoids/administration & dosage , Animals , Antigens, Neoplasm/metabolism , Cancer Vaccines/chemistry , Cell Line, Tumor , Cytotoxicity, Immunologic , Docetaxel , Humans , Hydrostatic Pressure , Immune System , Immunotherapy/methods , Interferon-gamma/metabolism , Interleukin-12/metabolism , Male , Mice , Mice, Inbred C57BL , Neoplasms, Experimental/drug therapy , Papillomavirus Infections/drug therapy , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Spleen/immunology , Toll-Like Receptor 9/metabolism
15.
J Leukoc Biol ; 95(5): 743-753, 2014 05.
Article in English | MEDLINE | ID: mdl-24389335

ABSTRACT

MDSCs represent one of the key players mediating immunosuppression. These cells accumulate in the TME, lymphoid organs, and blood during tumor growth. Their mobilization was also reported after CY therapy. DNMTi 5AC has been intensively studied as an antitumor agent. In this study, we examined, using two different murine tumor models, the modulatory effects of 5AC on TU-MDSCs and CY-MDSCs tumor growth and CY therapy. Indeed, the percentage of MDSCs in the TME and spleens of 5AC-treated mice bearing TRAMP-C2 or TC-1/A9 tumors was found decreased. The changes in the MDSC percentage were accompanied by a decrease in the Arg-1 gene expression, both in the TME and spleens. CY treatment of the tumors resulted in additional MDSC accumulation in the TME and spleens. This accumulation was subsequently inhibited by 5AC treatment. A combination of CY with 5AC led to the highest tumor growth inhibition. Furthermore, in vitro cultivation of spleen MDSCs in the presence of 5AC reduced the percentage of MDSCs. This reduction was associated with an increased percentage of CD11c+ and CD86+/MHCII+ cells. The observed modulatory effect on MDSCs correlated with a reduction of the Arg-1 gene expression, VEGF production, and loss of suppressive capacity. Similar, albeit weaker effects were observed when MDSCs from the spleens of tumor-bearing animals were cultivated with 5AC. Our findings indicate that beside the direct antitumor effect, 5AC can reduce the percentage of MDSCs accumulating in the TME and spleens during tumor growth and CY chemotherapy, which can be beneficial for the outcome of cancer therapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Myeloid-Derived Suppressor Cells/immunology , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/immunology , Animals , Azacitidine/pharmacology , Cell Line, Tumor , Cyclophosphamide/pharmacology , Male , Mice , Myeloid-Derived Suppressor Cells/pathology , Neoplasms, Experimental/pathology
16.
J Immunother ; 35(5): 374-84, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22576342

ABSTRACT

Myeloid-derived suppressor cells (MDSC) play an important role in tumor escape from antitumor immunity. MDSC accumulate in the lymphoid organs and blood during tumor growth and their mobilization was also reported after cyclophosphamide (CY) administration. In this communication, spleen MDSC accumulating after CY therapy (CY-MDSC) were compared with those expanded in mice bearing human papilloma viruses 16-associated TC-1 carcinoma (TU-MDSC). Although both CY-MDSC and TU-MDSC accelerated growth of TC-1 tumors in vivo, their phenotype and immunosuppressive function differed. CY-MDSC consisted of higher percentage of monocyte-like subpopulation and this was accompanied by lower relative expression of immunosuppressive genes and lower suppression of T-cell proliferation. After interferon-γ stimulation, the expression of immunosuppressive genes increased, but the suppressive ability of CY-MDSC did not reach that of TU-MDSC. The phenotype and function of MDSC obtained from mice bearing TC-1 tumors treated with CY was, in general, found to lie between CY-MDSC and TU-MDSC. After in vitro cultivation of MDSC in the presence of interleukin 12 (IL-12), the percentage of CD11b+/Gr-1+ cells decreased and was accompanied by an increase in the percentage of CD86+/MHCII+ cells. The strongest modulatory effect was noticed in the group of CY-MDSC. The susceptibility of CY-MDSC to all-trans-retinoic acid (ATRA) was also evaluated. In vitro cultivation with ATRA resulted in MDSC differentiation, and ATRA inhibited MDSC accumulation induced by CY administration. Our findings identified differences between CY-MDSC and TU-MDSC and supported the rationale for utilization of ATRA or IL-12 to alter MDSC accumulation after CY chemotherapy with the aim to improve its antitumor effect.


Subject(s)
Antineoplastic Agents/pharmacology , Cyclophosphamide/pharmacology , Myeloid Cells/immunology , Neoplasms, Experimental/immunology , Animals , B7-2 Antigen/biosynthesis , B7-2 Antigen/immunology , CD11b Antigen/biosynthesis , CD11b Antigen/immunology , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic , Human papillomavirus 16/immunology , Humans , Interferon-gamma/pharmacology , Interleukin-12/pharmacology , Lymphocyte Activation , Male , Mice , Mice, Inbred C57BL , Myeloid Cells/drug effects , Myeloid Cells/pathology , Neoplasm Transplantation , Neoplasms, Experimental/pathology , Neoplasms, Experimental/virology , Spleen/drug effects , Spleen/immunology , Spleen/pathology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Tretinoin/pharmacology
17.
Oncol Rep ; 25(6): 1683-9, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21424130

ABSTRACT

Genetically modified tumour cells producing cytokines such as interleukin 12 (IL-12) are potent activators of the antitumour immune responses and represent a promising therapeutic modality when combined with chemotherapy. The objective of this study was to examine whether IL-12-producing cellular vaccines can augment chemotherapy of human papilloma virus (HPV) 16-associated murine tumours with the cytostatic agent gemcitabine (GEM). We found that peritumoral administration of IL-12-producing tumour vaccines enhanced the effect of cytoreductive therapy with GEM both in non-metastasizing murine carcinoma TC-1 and in metastasizing murine carcinoma MK16. The percentage of mice with MK16 metastases and the number of lung metastatic nodules was substantially decreased. In another clinically relevant setting, surgical minimal residual tumour disease, the administration of IL-12-producing tumour vaccine and GEM after the MK16 tumour surgery reduced the percentage of mice with tumour recurrences; similarly, the percentage of metastasis-bearing mice and the number of metastatic nodules was decreased. Tumour inhibitory effects exerted by GEM plus IL-12 were associated with high production of interferon-γ (IFNγ) by splenocytes. Our results suggest that the IL-12-producing vaccine can enhance the effect of GEM chemotherapy in some HPV16-associated murine tumour models.


Subject(s)
Antineoplastic Agents/pharmacology , Cancer Vaccines/pharmacology , Deoxycytidine/analogs & derivatives , Genetic Therapy/methods , Interleukin-12/biosynthesis , Neoplasms, Experimental/drug therapy , Animals , Cell Separation , Combined Modality Therapy , Deoxycytidine/pharmacology , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Human papillomavirus 16 , Male , Mice , Mice, Inbred C57BL , Neoplasms, Experimental/virology , Papillomavirus Infections/complications , Gemcitabine
18.
Int J Cancer ; 126(12): 2997-3004, 2010 Jun 15.
Article in English | MEDLINE | ID: mdl-19739073

ABSTRACT

Natural killer T (NKT) cells are potent modulators of antitumor immunity. Their protective effects can be achieved upon their activation by glycolipid ligands presented in the context of the CD1d molecule. These CD1d-binding glycolipid antigens have been described as potent therapeutic agents against tumors, infections, as well as autoimmune diseases. Immunoregulatory and therapeutic effects of glycolipid ligands depend on their structure and modes of administration. Therefore, more studies are needed for optimization of the particular therapeutic settings. This study was focused on the tumor-inhibitory effects of 12 carbon acyl chain beta-galactosyl ceramide (C12 beta-D-Galactosyl Ceramide; beta-GalCer(C12)) on the growth of human papillomavirus type 16 (HPV16)-associated neoplasms transplanted in syngeneic mice. Treatment of tumor-bearing mice with beta-GalCer(C12) 3-14 days after tumor cell transplantation significantly inhibited the growth of the major histocompatibility complex (MHC) Class I-positive (TC-1), as well as MHC Class I-deficient (TC-1/A9) HPV16-associated tumors. Moreover, administration of beta-GalCer(C12) after surgical removal of TC-1 tumors inhibited the growth of tumor recurrences. Similar results were obtained in the treatment of tumors after chemotherapy. beta-GalCer(C12) treatment turned out to be also synergistic with immunotherapy based on administration of IL-12-producing cellular vaccines. These results suggest that beta-GalCer(C12), whose antitumor effects have so far not been studied in detail, can be effective for the treatment of minimal residual tumor disease as well as an adjuvant for cancer immunotherapy.


Subject(s)
Ceramides/pharmacology , Monosaccharides/pharmacology , Neoplasm Recurrence, Local/pathology , Neoplasm Recurrence, Local/prevention & control , Neoplasm, Residual/drug therapy , Neoplasm, Residual/surgery , Papillomavirus Infections/pathology , Papillomavirus Infections/prevention & control , Animals , Human papillomavirus 16/isolation & purification , Humans , Immunotherapy , Male , Mice , Mice, Inbred C57BL , Neoplasm Recurrence, Local/immunology , Neoplasm, Residual/virology , Papillomavirus Infections/immunology , Tumor Cells, Cultured/transplantation
19.
Int J Oncol ; 26(2): 521-7, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15645139

ABSTRACT

Malignant transformation of somatic cells followed by selection of the transformed cell populations can give rise to tumours that display an immune escape phenotype, MHC class I deficient neoplasms. Experiments were designed to examine whether the immune escape phenotype of HPV16-associated tumours is stable or whether the MHC class I expression can change during tumour progression and therapy. It has been found that temporary growth of MHC class I- tumour MK16/1/IIIABC in syngeneic mice can lead to up-regulation of the low MHC class I expression, both in the subcutaneous tumour inocula and in their lung metastases. Mimicking this process in vitro by co-cultivation of tumour and spleen cell populations revealed that the spleen cells produce IFNgamma, which upregulates MHC class I expression on the MK16/1/IIIABC cells as well as their sensitivity to T cell-mediated cytolysis (CTLs). The up-regulation could be prevented by admixture of anti-IFNgamma antibody to the tumour/spleen cell mixtures. Similar up-regulation of the MHC class I expression was observed in HPV16-associated tumour cell lines, MK16/1/IIIABC, MK16/MET/M1, TC-1, TC-1/A9 and TC-1/P3C10 grown in vitro in the presence of IFNgamma. The up-regulation was found to be IFNgamma dose-dependent and the level of the MHC class I expression required for in vitro cytolysis of the tumour cells by CTLs could be characterized in cytofluorometry with anti-H-2 antibody. After removal of the IFNgamma from the cultivation medium or after injection of the IFNgamma-treated cells into syngeneic mice the MHC class I expression gradually dropped back to the original level or to the level observed on the tumours growing in vivo. These findings indicate that the immune escape phenotype of at least some HPV16-associated tumours is not stable and that up-regulation of the MHC class I expression can occur in vivo during progression of the MHC I- tumours, apparently due to production of IFNgamma by the immune cells in the tumour microenviroment and its vicinity. In vitro irradiation of HPV16-associated MHC class I-deficient tumour cell lines MK16/MET/M1 and TC-1/P3C10 with a dose of 150 Gy up-regulated their MHC class I expression. Similarly, substantial up-regulation of the MHC class I expression was observed in TC-1/A9 tumour recurrences after surgery. The up-regulation observed in the recurrences after surgery or after irradiation has reached the level required for in vitro cytolysis of the tumour cells by CTLs. If confirmed also with other tumour types and in human tumour systems, the up-regulation of MHC class I molecule expression during radiotherapy and in tumour recurrences after surgery may have important implications in the development of immunotherapeutic strategies.


Subject(s)
Gene Expression Regulation, Neoplastic , Histocompatibility Antigens Class I/biosynthesis , Immunotherapy/methods , Neoplasms/immunology , Neoplasms/virology , Oncogene Proteins, Viral/biosynthesis , Papillomaviridae/metabolism , Papillomavirus Infections/immunology , Animals , Cell Line, Tumor , Chromium Radioisotopes , Coculture Techniques , Disease Progression , Flow Cytometry , Genes, MHC Class I , Interferon-gamma/biosynthesis , Male , Mice , Mice, Inbred C57BL , Neoplasms, Experimental , Phenotype , Recurrence , Spleen/metabolism , T-Lymphocytes, Cytotoxic/metabolism , Transfection , Up-Regulation
20.
Int J Oncol ; 25(4): 1165-70, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15375569

ABSTRACT

Dendritic cell (DC)-based vaccines are being intensively investigated for the treatment of a variety of human neoplasms. However, little attention has until now been paid to the use of DC-based vaccines for immunotherapy of tumour residua after surgery. In this communication, an animal model mimicking human HPV16-associated neoplasms was employed to examine the effect of DC-based vaccines for the treatment of surgical minimal residual tumour disease. Mice were subcutaneously inoculated with syngeneic TC-1 tumour cells of HPV16 origin. When the tumours reached approximately 1 cm in diameter, they were surgically removed and the operated mice were injected into the site of the operation with bone marrow-derived DC, which were either pulsed with TC-1 cell lysates or co-cultured with irradiated TC-1 cells. It has been found that the growth of TC-1 tumour recurrences in the mice treated with these vaccines was substantially suppressed, as compared to the operated-only controls. The phenotypic analysis of the spleen cells has shown that the percentage of CD3+ cells was diminished in the operated-only and vaccinated mice carrying recurrent tumours, in comparison with healthy control mice and with operated tumour-free mice. Moreover, accumulation of immature myeloid cells (CD11b+/Gr-1+) was observed in spleens of the tumour-bearing mice. These findings indicate that the immune system of the tumour-bearing individuals was compromised, as compared to that of normal individuals or tumour regressors. To our knowledge, this is the first report that has demonstrated the positive effect of local administration of the DC-based, HPV16 E6/E7 oncoprotein-containing, tumour lysate-loaded vaccines in the treatment of surgical minimal residual tumour disease.


Subject(s)
Cancer Vaccines/immunology , Dendritic Cells/immunology , Neoplasms, Experimental/therapy , Oncogene Proteins, Viral/immunology , Papillomaviridae/immunology , Papillomavirus Infections/complications , Repressor Proteins/immunology , Animals , Immunophenotyping , Male , Mice , Mice, Inbred C57BL , Neoplasm, Residual , Neoplasms, Experimental/immunology , Papillomavirus E7 Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...